Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Curr Protoc ; 3(11): e925, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37934124

ABSTRACT

The MHC II-EGFP knock-in mouse model enables us to visualize and track MHC-II-expressing cells in vivo by expressing enhanced green fluorescent protein (EGFP) fused to the MHC class II molecule under the MHC II beta chain promoter. Using this model, we can easily identify MHC-II-expressing cells, including dendritic cells, B cells, macrophages, and ILC3s, which play a key role as antigen-presenting cells (APCs) for CD4+ T cells. In addition, we can also precisely identify and analyze APC-containing tissues and organs. Even after fixation, EGFP retains its fluorescence, so this model is suitable for immunofluorescence studies, facilitating an unbiased characterization of the histological context, especially with techniques such as light-sheet fluorescence microscopy. Furthermore, the MHC II-EGFP knock-in mouse model is valuable for studying the molecular mechanisms of MHC II gene regulation and expression by making it possible to correlate MHC II expression (MHC II-EGFP) with surface fraction through antibody detection, thereby shedding light on the intricate regulation of MHC II expression. Overall, this model is an essential asset for quantitative and systems immunological research, providing insights into immune cell dynamics and localization, with a tool for precise cell identification and with the ability to study MHC II gene regulation, thus furthering the understanding of immune responses and underlying mechanisms © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Characterization of antigen-specific MHC II loading compartment tubulation toward the immunological synapse Basic Protocol 2: Characterization of overall versus surface MHC II expression Basic Protocol 3: Identification and preparation of the lymphoid organs Basic Protocol 4: Quantification of APC content in lymphoid organs by fluorescence stereomicroscopy Basic Protocol 5: Quantification and measurement of intestinal lymphoid tissue by light-sheet fluorescence stereomicroscopy Basic Protocol 6: Visualization of corneal APCs Basic Protocol 7: Quantification of MHC II+ cells in maternal milk by flow cytometry Support Protocol 1: Cell surface staining and flow cytometry analysis of spleen mononuclear cells.


Subject(s)
Antigen-Presenting Cells , B-Lymphocytes , Animals , Mice , Green Fluorescent Proteins , Cell Membrane , Disease Models, Animal
3.
Immunol Lett ; 251-252: 75-85, 2022 12.
Article in English | MEDLINE | ID: mdl-36332824

ABSTRACT

Immunology is a rapidly evolving field of research with sophisticated models and methods. However, detailed data on total immune cell counts and population distributions remain surprisingly scarce. Nevertheless, recently established quantitative approaches could help us understand the overall complexity of the immune system. Here, we studied a major histocompatibility complexclass II - enhanced green fluorescent protein knock-in mouse model to precisely identify and manipulate lymphoid structures. By combining flow cytometry with light sheet microscopy, we quantified MHC II+ populations of the small intestine and associated individual mesenteric lymph nodes, with 36.7 × 106 cells in lamina propria, 3.0 × 105 cells in scattered lymphoid tissue and 1.1 × 106 cells in Peyer's patches. In addition to these whole-organ cell counts, we assessed approximately 1 × 106 total villi in the small intestine and 450 scattered lymphoid tissue follicles. By direct noninvasive microscopic observation of a naturally fully translucent mouse organ, the cornea, we quantified 12 ± 4 and 35 ± 7 cells/mm2 Langerhans- and macrophage-like populations, respectively. Ultimately, our findings show that flow cytometry with quantitative imaging data analysis enables us to avoid methodological discrepancies while gaining new insights into the relevance of organ-specific quantitative approaches for immunology.


Subject(s)
Lymphoid Tissue , Peyer's Patches , Animals , Mice , Intestinal Mucosa , Intestine, Small , Lymph Nodes , Histocompatibility Antigens Class II/immunology
4.
ChemMedChem ; 17(21): e202200385, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36115047

ABSTRACT

Ketoconazole (KTZ) is an imidazole drug applied topically to treat numerous skin infections. However, as a systemic antifungal, KTZ' efficacy and safety no longer justify its use as a first-line treatment. Azole conjugates often display higher solubility and better antifungal activities than their parent azoles. Accordingly, we aimed at developing suitable linkers for clickable azole conjugation with a second antifungal molecule, and targeted drug delivery towards improving antifungal activity. For its low price and high availability, we selected KTZ as a molecular scaffold to introduce such chemical modifications. We prepared a series of piperazine-modified KTZ derivatives and we evaluated their in vitro antifungal and antitrypanosomal activity against fourteen strains of pathogenic fungi and two strains of Trypanosoma parasites. Several compounds were more effective against the pathogens than KTZ. Compound 5 was 24 times more potent against Aspergillus flavus and 8 times more potent against A. fumigatus than KTZ, with similarly low cytotoxicity to HEK cells up to 100 µM. Derivative 6 had 9- and 7-fold higher activity against T. brucei gambiense and T. brucei brucei than KTZ, respectively, and inhibited trypanosoma growth at single micromolar EC50 values. Combined, our findings will foster further research of piperazine-modified KTZs as promising antifungal and antiparasitic drugs towards enhancing the properties of both KTZ and other azole derivatives.


Subject(s)
Antifungal Agents , Ketoconazole , Ketoconazole/pharmacology , Ketoconazole/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Azoles
5.
Nat Commun ; 13(1): 5022, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028489

ABSTRACT

Signaling by the human C-type lectin-like receptor, natural killer (NK) cell inhibitory receptor NKR-P1, has a critical role in many immune-related diseases and cancer. C-type lectin-like receptors have weak affinities to their ligands; therefore, setting up a comprehensive model of NKR-P1-LLT1 interactions that considers the natural state of the receptor on the cell surface is necessary to understand its functions. Here we report the crystal structures of the NKR-P1 and NKR-P1:LLT1 complexes, which provides evidence that NKR-P1 forms homodimers in an unexpected arrangement to enable LLT1 binding in two modes, bridging two LLT1 molecules. These interaction clusters are suggestive of an inhibitory immune synapse. By observing the formation of these clusters in solution using SEC-SAXS analysis, by dSTORM super-resolution microscopy on the cell surface, and by following their role in receptor signaling with freshly isolated NK cells, we show that only the ligation of both LLT1 binding interfaces leads to effective NKR-P1 inhibitory signaling. In summary, our findings collectively support a model of NKR-P1:LLT1 clustering, which allows the interacting proteins to overcome weak ligand-receptor affinity and to trigger signal transduction upon cellular contact in the immune synapse.


Subject(s)
Killer Cells, Natural , Receptors, Cell Surface , Antigens, Surface , Cluster Analysis , Humans , Lectins, C-Type , Ligands , NK Cell Lectin-Like Receptor Subfamily B , Scattering, Small Angle , Synapses , X-Ray Diffraction
6.
Toxins (Basel) ; 11(8)2019 07 25.
Article in English | MEDLINE | ID: mdl-31349616

ABSTRACT

Ergot, fungal genus Claviceps, are worldwide distributed grass pathogens known for their production of toxic ergot alkaloids (EAs) and the great agricultural impact they have on both cereal crop and farm animal production. EAs are traditionally considered as the only factor responsible for ergot toxicity. Using broad sampling covering 13 ergot species infecting wild or agricultural grasses (including cereals) across Europe, USA, New Zealand, and South Africa we showed that the content of ergochrome pigments were comparable to the content of EAs in sclerotia. While secalonic acids A-C (SAs), the main ergot ergochromes (ECs), are well known toxins, our study is the first to address the question about their contribution to overall ergot toxicity. Based on our and published data, the importance of SAs in acute intoxication seems to be negligible, but the effect of chronic exposure needs to be evaluated. Nevertheless, they have biological activities at doses corresponding to quantities found in natural conditions. Our study highlights the need for a re-evaluation of ergot toxicity mechanisms and further studies of SAs' impact on livestock production and food safety.


Subject(s)
Claviceps/chemistry , Ergot Alkaloids/toxicity , Mycotoxins/toxicity , Xanthenes/toxicity , Apoptosis/drug effects , Cell Survival/drug effects , Ergot Alkaloids/analysis , HeLa Cells , Humans , Jurkat Cells , Mitochondria/drug effects , Mycotoxins/analysis , Mycotoxins/pharmacology , Xanthenes/analysis
7.
J Proteomics ; 196: 162-172, 2019 03 30.
Article in English | MEDLINE | ID: mdl-30414947

ABSTRACT

The cytotoxicity of mouse natural killer (NK) cells in response to pathological changes in target cells is regulated via the Nkrp1b receptor. Here, we characterized the Nkrp1b structure and structural features (stalk, loop, and oligomerization state) that affect its interactions. To study the Nkrp1b protein structure and the functional importance of its stalk, two Nkrp1b protein variants differing by the presence of the stalk were prepared. These variants were studied using a combination of structural mass spectrometry approaches with computational modeling to derive structural models. In addition, information about biological activity and localization in mammalian cells was acquired using scanning microscopy techniques and western blotting. Based on these methods, we obtained the structure of Nkrp1b ectodomain in its monomeric and dimeric conformations, identified the dimerization interface, and determined disulfide connections within the molecule. We found that Nkrp1b occurs as a mixture of monomers and homodimers, both in vitro and in vivo. SIGNIFICANCE: Despite the long-standing assumption that Nkrp1 proteins are homodimers connected by disulfide bonds in the stalk region, our data showed that both Nkrp1b protein variants form monomers and homodimers irrespective of the presence of the stalk. We demonstrated that the stalk is not crucial for protein dimerization or ligand binding and that Nkrp1b interacts with its natural ligands only in its monomeric conformation; therefore, dimers may have another regulatory function. Using a unique combination of computational, biochemical, and biological methods, we revealed the structural conformation and behavior of Nkrp1b in its native state. In addition, it is a first report utilizing the intermolecular chemical cross-linking of light- and heavy-labeled protein chains together with ion mobility-mass spectrometry to design the structural models of protein homodimers.


Subject(s)
Models, Molecular , NK Cell Lectin-Like Receptor Subfamily B/chemistry , Protein Multimerization , Proteomics , Animals , Mice , Mice, Inbred BALB C , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Protein Structure, Secondary , Structure-Activity Relationship
8.
J Allergy Clin Immunol ; 142(6): 1947-1955, 2018 12.
Article in English | MEDLINE | ID: mdl-29432774

ABSTRACT

BACKGROUND: Proline-serine-threonine phosphatase interacting protein 1 (PSTPIP1) is a cytosolic adaptor protein involved with T-cell activation, differentiation, and migration. On cognate T-cell contact, PSTPIP1 is recruited to surface-expressed CD2, where it regulates F-actin remodeling. An immune synapse (IS) is thereby rapidly formed, consisting of T-cell receptor clusters surrounded by a ring of adhesion molecules, including CD2. OBJECTIVE: From genetic screening of patients with primary immunodeficiencies, we identified 2 mutations in PSTPIP1, R228C and T274M, which we further characterized in the primary patients' T cells. METHODS: F-actin dynamics were assessed in primary T cells from the patients and control subjects by using fluorescence-activated cell sorting. HEK293T and Jurkat cells were transfected with R228C, T274M, and wild-type PSTPIP1 to visualize F-actin in IS formation. CD2-PSTPIP1 association was quantified through immunoprecipitation assays. RESULTS: The patients presented with immunodeficiency without signs of autoinflammation. The patient with the R228C mutation had expansion of mostly naive phenotype T cells and few memory T cells; the patient with the T274M mutation had 75% reduction in CD4 T cells that were predominantly of the memory subset. We observed F-actin polymerization defects in T cells from both patients with PSTPIP1, most notably the patient with the T274M mutation. Capping of CD2-containing membrane microdomains was disrupted. Analysis of IS formation using Jurkat T-cell transfectants revealed a reduction in F-actin accumulation at the IS, again especially in cells from the patient with the T274M PSTPIP1 mutation. T cells from the patient with the T274M mutation migrated spontaneously at increased speed, as assessed in a 3-dimensional collagen matrix, whereas T-cell receptor cross-linking induced a significantly diminished calcium flux. CONCLUSIONS: We propose that PSTPIP1 T-cell differentiation defects are caused by defective control of F-actin polymerization. A preactivated polymerized F-actin status, as seen in T cells from patients with the PSTPIP1 T274M mutation, appears particularly damaging. PSTPIP1 controls IS formation and cell adhesion through its function as an orchestrator of the F-actin cytoskeleton.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/immunology , Common Variable Immunodeficiency , Cytoskeletal Proteins/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion , Cell Differentiation , Cell Movement , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/metabolism , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Mutation , T-Lymphocytes/physiology
9.
Oncotarget ; 8(61): 103137-103153, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262552

ABSTRACT

Abnormalities in cancer metabolism represent potential targets for cancer therapy. We have recently identified a natural compound Quambalarine B (QB), which inhibits proliferation of several leukemic cell lines followed by cell death. We have predicted ubiquinone binding sites of mitochondrial respiratory complexes as potential molecular targets of QB in leukemia cells. Hence, we tracked the effect of QB on leukemia metabolism by applying several omics and biochemical techniques. We have confirmed the inhibition of respiratory complexes by QB and found an increase in the intracellular AMP levels together with respiratory substrates. Inhibition of mitochondrial respiration by QB triggered reprogramming of leukemic cell metabolism involving disproportions in glycolytic flux, inhibition of proteins O-glycosylation, stimulation of glycine synthesis pathway, and pyruvate kinase activity, followed by an increase in pyruvate and a decrease in lactate levels. Inhibition of mitochondrial complex I by QB suppressed folate metabolism as determined by a decrease in formate production. We have also observed an increase in cellular levels of several amino acids except for aspartate, indicating the dependence of Jurkat (T-ALL) cells on aspartate synthesis. These results indicate blockade of mitochondrial complex I and II activity by QB and reduction in aspartate and folate metabolism as therapeutic targets in T-ALL cells. Anti-cancer activity of QB was also confirmed during in vivo studies, suggesting the therapeutic potential of this natural compound.

10.
Exp Cell Res ; 349(2): 273-281, 2016 Dec 10.
Article in English | MEDLINE | ID: mdl-27793648

ABSTRACT

The general mechanism underlying the tumor suppressor activity of the Hippo signaling pathway remains unclear. In this study, we explore the molecular mechanisms connecting the Hippo signaling pathway with glucose metabolism. We have found that two key regulators of glycolysis, C-MYC and GLUT1, are targets of the Hippo signaling pathway in human leukemia cells. Our results revealed that activation of MST1 by the natural compound shikonin inhibited the expression of GLUT1 and C-MYC. Furthermore, RNAi experiments confirmed the regulation of GLUT1 and C-MYC expression via the MST1-YAP1-TEAD1 axis. Surprisingly, YAP1 was found to positively regulate C-MYC mRNA levels in complex with TEAD1, while it negatively regulates C-MYC levels in cooperation with MST1. Hence, YAP1 serves as a rheostat for C-MYC, which is regulated by MST1. In addition, depletion of MST1 stimulates lactate production, whereas the specific depletion of TEAD1 has an opposite effect. The inhibition of lactate production and cellular proliferation induced by shikonin also depends on the Hippo pathway activity. Finally, a bioinformatic analysis revealed conserved TEAD-binding motifs in the C-MYC and GLUT1 promoters providing another molecular data supporting our observations. In summary, regulation of glucose metabolism could serve as a new tumor suppressor mechanism orchestrated by the Hippo signaling pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Genes, myc/drug effects , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/drug effects , Apoptosis/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/drug effects , Glucose Transporter Type 1/metabolism , Hepatocyte Growth Factor , Humans , Nuclear Proteins/drug effects , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Proto-Oncogene Proteins/drug effects , Signal Transduction/physiology , TEA Domain Transcription Factors , Transcription Factors/drug effects , YAP-Signaling Proteins
11.
J Nat Prod ; 79(9): 2304-14, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27571379

ABSTRACT

Quambalarine B (QB) is a secondary metabolite produced by the basidiomycete Quambalaria cyanescens with potential anticancer activity. Here we report that QB at low micromolar concentration inhibits proliferation of several model leukemic cell lines (Jurkat, NALM6, and REH), whereas higher concentrations induce cell death. By contrast, the effect of QB on primary leukocytes (peripheral blood mononuclear cells) is significantly milder with lower toxicity and cytostatic activity. Moreover, QB inhibited expression of the C-MYC oncoprotein and mRNA expression of its target genes, LDHA, PKM2, and GLS. Finally, QB blocked the phosphorylation of P70S6K, a downstream effector kinase in mTOR signaling that regulates translation of C-MYC. This observation could explain the molecular mechanism behind the antiproliferative and cytotoxic effects of QB on leukemic cells. Altogether, our results establish QB as a promising molecule in anticancer treatment.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Basidiomycota/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Antineoplastic Agents/blood , Antineoplastic Agents/isolation & purification , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Jurkat Cells/drug effects , Leukocytes, Mononuclear/drug effects , Molecular Structure , Naphthoquinones/blood , Naphthoquinones/chemical synthesis , Naphthoquinones/isolation & purification , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa , Signal Transduction/physiology , TOR Serine-Threonine Kinases
12.
Molecules ; 20(2): 3463-78, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25690298

ABSTRACT

The C-type lectin-like receptors include the Nkrp1 protein family that regulates the activity of natural killer (NK) cells. Rat Nkrp1a was reported to bind monosaccharide moieties in a Ca2+-dependent manner in preference order of GalNac > GlcNAc >> Fuc >> Gal > Man. These findings established for rat Nkrp1a have been extrapolated to all additional Nkrp1 receptors and have been supported by numerous studies over the past two decades. However, since 1996 there has been controversy and another article showed lack of interactions with saccharides in 1999. Nevertheless, several high affinity saccharide ligands were synthesized in order to utilize their potential in antitumor therapy. Subsequently, protein ligands were introduced as specific binders for Nkrp1 proteins and three dimensional models of receptor/protein ligand interaction were derived from crystallographic data. Finally, for at least some members of the NK cell C-type lectin-like proteins, the "sweet story" was impaired by two reports in recent years. It has been shown that the rat Nkrp1a and CD69 do not bind saccharide ligands such as GlcNAc, GalNAc, chitotetraose and saccharide derivatives (GlcNAc-PAMAM) do not directly and specifically influence cytotoxic activity of NK cells as it was previously described.


Subject(s)
Antigens, CD , Antigens, Differentiation, T-Lymphocyte , Killer Cells, Natural , Lectins, C-Type , NK Cell Lectin-Like Receptor Subfamily B , Oligosaccharides , Animals , Antigens, CD/chemistry , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/chemistry , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Humans , Killer Cells, Natural/chemistry , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , NK Cell Lectin-Like Receptor Subfamily B/chemistry , NK Cell Lectin-Like Receptor Subfamily B/immunology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Oligosaccharides/chemistry , Oligosaccharides/immunology , Oligosaccharides/metabolism , Protein Structure, Tertiary , Rats
13.
Nanoscale ; 6(20): 11712-21, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25037888

ABSTRACT

Nanodiamonds (NDs) are versatile nanoparticles that are currently being investigated for a variety of applications in drug delivery, biomedical imaging and nanoscale sensing. Although initial studies indicate that these small gems are biocompatible, there is a great deal of variability in synthesis methods and surface functionalization that has yet to be evaluated. Here we present a comprehensive analysis of the cellular compatibility of an array of nanodiamond subtypes and surface functionalization strategies. These results demonstrate that NDs are well tolerated by multiple cell types at both functional and gene expression levels. In addition, ND-mediated delivery of daunorubicin is less toxic to multiple cell types than treatment with daunorubicin alone, thus demonstrating the ability of the ND agent to improve drug tolerance and decrease therapeutic toxicity. Overall, the results here indicate that ND biocompatibility serves as a promising foundation for continued preclinical investigation.


Subject(s)
Biocompatible Materials/chemistry , Nanodiamonds/chemistry , Nanotechnology/methods , Amines/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Colloids/chemistry , Daunorubicin/chemistry , Drug Delivery Systems , Gene Expression Profiling , HeLa Cells , Hep G2 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Microscopy, Fluorescence , Particle Size , Surface Properties , Water/chemistry
14.
Immunol Lett ; 156(1-2): 110-7, 2013.
Article in English | MEDLINE | ID: mdl-24076118

ABSTRACT

Recognition of glycosylation patterns is one of the basic features of innate immunity. Ability of C-type lectin-like receptors such as NKR-P1 to bind saccharide moieties has become recently a controversial issue. In the present study, binding assay with soluble fluorescently labeled recombinant rat NKR-P1A and mouse NKR-P1C proteins revealed apparently no affinity to the various neoglycoproteins. Lack of functional linkage between NKR-P1 and previously described saccharide binder was supported by the fact, that synthetic N-acetyl-D-glucosamine octabranched dendrimer on polyamidoamine scaffold (GN8P) did not change gene expression of NKR-P1 isoforms in C57BL/6 and BALB/c mice divergent in the NK gene complex (both in vitro and in vivo). Surprisingly, N-acetyl-D-glucosamine-coated tetrabranched polyamido-amine dendrimer specifically binds to NKT cells and macrophages but not to NK cells (consistently with changes in cytokine patterns). Despite the fact that GN8P has been tested as an immunomodulator in anti-cancer treatment animal models for many years, surprisingly no changes in cytokine profiles in serum relevant to anti-cancer responses using B16F10 and CT26 harboring mouse strains C57BL/6 and BALB/c are observed. Our results indicate possible indirect involvement of NK cells in GN8P mediated immune responses.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Oligosaccharides/immunology , Acetylglucosamine/immunology , Acetylglucosamine/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Dendrimers/metabolism , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Glycoconjugates/immunology , Glycoconjugates/metabolism , Glycoconjugates/pharmacology , Interferon-gamma/blood , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/genetics , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Oligosaccharides/metabolism , Polyamines/immunology , Polyamines/metabolism , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
Int Immunopharmacol ; 11(8): 955-61, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21349367

ABSTRACT

N-acetyl-D-glucosamine-coated polyamidoamine dendrimer (GN8P), exerting high binding affinity to rodent recombinant NKR-P1A and NKR-P1C activating proteins, was shown previously to delay the development of rat colorectal carcinoma as well as mouse B16F10 melanoma, and to potentiate antigen-specific antibody formation in healthy C57BL/6 mice via NK cell stimulation. In this study, we investigated whether GN8P also modulates tumor-specific B cell responses. Serum anti-B16F10 melanoma IgG levels, IgG2a mRNA expression, antibody dependent cell-mediated cytotoxicity (ADCC), and counts of plasma as well as antigen presenting B cells were evaluated in tumor-bearing C57BL/6 mice treated with GN8P and in respective controls. To reveal the mechanism of GN8P effects, the synthesis of interferon-gamma (IFN-γ) and interleukin-4 (IL-4), cytokines involved in regulation of immunoglobulin class switch, was determined. The GN8P treatment significantly elevated IgG, and particularly IgG2a, response against B16F10 melanoma, which led to augmented ADCC reaction. The significant increase in production of IFN-γ, which is known to support IgG2a secretion, was observed solely in NK1.1 expressing cell populations, predominantly in NK cells. Moreover, GN8P raised the number of plasma cells, and promoted antigen presenting capacity of I-A/I-E-positive B lymphocytes by up-regulation of their CD80 and CD86 co-stimulatory molecule expression. These results indicate that GN8P-induced enhancement of tumor-specific antibody formation is triggered by NK cell activation, and contributes to complexity of anticancer immune response involving lectin-saccharide interaction.


Subject(s)
Acetylglucosamine/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Dendrimers/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Acetylglucosamine/chemistry , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/metabolism , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-2 Antigen/biosynthesis , B7-2 Antigen/genetics , Cell Line, Tumor , Dendrimers/chemistry , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Killer Cells, Natural/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Up-Regulation/drug effects
16.
J Immunotoxicol ; 6(4): 217-26, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19908940

ABSTRACT

The intestinal environment is considered to play an important role both in colorectal tumor development and in the evolution and modulation of mucosal immunity. Studies in animals reared in germ-free (GF, without any intestinal microflora) versus conventional (CV, with regular microflora in bowel) conditions can aid in clarifying the influence of bacteria on carcinogenesis and anti-cancer immune responses in situ. The lower incidence of colon cancers and better immunological parameters in GF animals versus CV ones after chemically-induced carcinogenesis raises questions about specific characteristics of the immunological networks in each respective condition. Different levels of tolerance/regulatory mechanisms in the GF versus CV animals may influence the development of immune responses not only at the level of mucosal, but also at the systemic, immunity. We hypothesize that GF animals can better recognize and respond to evolving neoplasias in the bowel as a consequence of their less-tolerogenic immunity (i.e., due to their more limited exposure to antigens to become tolerated against at the intestinal level). In this paper, we review the role of bacteria in modulating gut environment and mucosal immunity, their importance in cancer development, and aspects of immune regulation (both at local and systemic level) that can be modified by bacterial microflora. Lastly, the use of GF animals in comparison with conventionally-raised animals is proposed as a suitable and potent model for understanding the inflammatory network and its effect on cancer immunity especially during colorectal cancer development.


Subject(s)
Adenocarcinoma/immunology , Colon/immunology , Colorectal Neoplasms/immunology , Germ-Free Life/immunology , Immunity, Innate , Adenocarcinoma/microbiology , Animals , Colon/microbiology , Colorectal Neoplasms/microbiology , Disease Models, Animal , Germ-Free Life/drug effects , Humans , Immune Tolerance , Immunity, Mucosal/immunology , Killer Cells, Natural/immunology , Mice , Rats , Receptor Cross-Talk/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...